Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 16.224
Filter
1.
Methods Mol Biol ; 2807: 175-194, 2024.
Article in English | MEDLINE | ID: mdl-38743229

ABSTRACT

Retroviruses must overcome cellular restrictions to the nucleocytoplasmic export of viral mRNAs that retain introns in order to complete their replication cycle. HIV accomplishes this using a system comprised of a trans-acting viral protein, Rev, and a cis-acting RNA secondary structure in the viral genome, the Rev-Response Element (RRE). HIV primary isolates differ with respect to the sequence and functional activity of the Rev-RRE system. Here, we describe a high throughput assay system for analyzing Rev-RRE functional activity using packageable viral vectors.


Subject(s)
RNA, Viral , Response Elements , rev Gene Products, Human Immunodeficiency Virus , Humans , rev Gene Products, Human Immunodeficiency Virus/genetics , rev Gene Products, Human Immunodeficiency Virus/metabolism , Response Elements/genetics , RNA, Viral/genetics , HIV-1/genetics , HIV-1/physiology , Gene Expression Regulation, Viral , Virus Replication/genetics , Genetic Vectors/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
2.
Methods Mol Biol ; 2808: 35-56, 2024.
Article in English | MEDLINE | ID: mdl-38743361

ABSTRACT

Mononegaviruses are promising tools as oncolytic and transgene vectors for gene therapy and regenerative medicine. However, when mononegaviruses are used for therapeutic applications, the viral activity must be strictly controlled due to concerns about toxicity and severe side effects. With this technology, mononegavirus vectors can be grown where they are intended and can be easily removed when they are no longer needed. In particular, a photoswitch protein called Magnet (consisting of two magnet domains) is incorporated into the hinge region between the connector and methyltransferase domains of the mononegavirus polymerase protein (L protein) to disrupt the L protein functions. Blue light (470 ± 20 nm) irradiation causes the dimerization of the two magnet domains, and the L protein is restored to activity, allowing viral gene expression and virus replication. Since the magnet domains' dimerization is reversible, viral gene expression and replication cease when blue light irradiation is stopped.


Subject(s)
Gene Expression Regulation, Viral , Virus Replication , Virus Replication/genetics , Humans , Viral Proteins/genetics , Viral Proteins/metabolism , Light , Animals , Genetic Vectors/genetics
3.
Nat Commun ; 15(1): 3729, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702330

ABSTRACT

The unique virus-cell interaction in Epstein-Barr virus (EBV)-associated malignancies implies targeting the viral latent-lytic switch is a promising therapeutic strategy. However, the lack of specific and efficient therapeutic agents to induce lytic cycle in these cancers is a major challenge facing clinical implementation. We develop a synthetic transcriptional activator that specifically activates endogenous BZLF1 and efficiently induces lytic reactivation in EBV-positive cancer cells. A lipid nanoparticle encapsulating nucleoside-modified mRNA which encodes a BZLF1-specific transcriptional activator (mTZ3-LNP) is synthesized for EBV-targeted therapy. Compared with conventional chemical inducers, mTZ3-LNP more efficiently activates EBV lytic gene expression in EBV-associated epithelial cancers. Here we show the potency and safety of treatment with mTZ3-LNP to suppress tumor growth in EBV-positive cancer models. The combination of mTZ3-LNP and ganciclovir yields highly selective cytotoxic effects of mRNA-based lytic induction therapy against EBV-positive tumor cells, indicating the potential of mRNA nanomedicine in the treatment of EBV-associated epithelial cancers.


Subject(s)
Epstein-Barr Virus Infections , Herpesvirus 4, Human , Liposomes , Nanoparticles , Trans-Activators , Humans , Herpesvirus 4, Human/genetics , Trans-Activators/metabolism , Trans-Activators/genetics , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Infections/drug therapy , Animals , Nanoparticles/chemistry , Cell Line, Tumor , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , Virus Activation/drug effects , Xenograft Model Antitumor Assays , Gene Expression Regulation, Viral/drug effects , Mice, Nude , Female
4.
Molecules ; 29(9)2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38731543

ABSTRACT

Ribonuclease P (RNase P) complexed with an external guide sequence (EGS) represents a promising nucleic acid-based gene targeting approach for gene expression knock-down and modulation. The RNase P-EGS strategy is unique as an EGS can be designed to basepair any mRNA sequence and recruit intracellular RNase P for hydrolysis of the target mRNA. In this study, we provide the first direct evidence that the RNase P-based approach effectively blocks the gene expression and replication of herpes simplex virus 2 (HSV-2), the causative agent of genital herpes. We constructed EGSs to target the mRNA encoding HSV-2 single-stranded DNA binding protein ICP8, which is essential for viral DNA genome replication and growth. In HSV-2 infected cells expressing a functional EGS, ICP8 levels were reduced by 85%, and viral growth decreased by 3000 folds. On the contrary, ICP8 expression and viral growth exhibited no substantial differences between cells expressing no EGS and those expressing a disabled EGS with mutations precluding RNase P recognition. The anti-ICP8 EGS is specific in targeting ICP8 because it only affects ICP8 expression but does not affect the expression of the other viral immediate-early and early genes examined. This study shows the effective and specific anti-HSV-2 activity of the RNase P-EGS approach and demonstrates the potential of EGS RNAs for anti-HSV-2 applications.


Subject(s)
Gene Expression Regulation, Viral , Herpesvirus 2, Human , Virus Replication , Herpesvirus 2, Human/genetics , Herpesvirus 2, Human/physiology , Humans , Ribonuclease P/metabolism , Ribonuclease P/genetics , Animals , Viral Proteins/genetics , Viral Proteins/metabolism , Chlorocebus aethiops , RNA, Messenger/genetics , RNA, Messenger/metabolism , Vero Cells , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , DNA-Binding Proteins
5.
Viruses ; 16(4)2024 Mar 25.
Article in English | MEDLINE | ID: mdl-38675845

ABSTRACT

Schlafen (SLFN) is a family of proteins upregulated by type I interferons with a regulatory role in translation. Intriguingly, SLFN14 associates with the ribosome and can degrade rRNA, tRNA, and mRNA in vitro, but a role in translation is still unknown. Ribosomes are important regulatory hubs during translation elongation of mRNAs rich in rare codons. Therefore, we evaluated the potential role of SLFN14 in the expression of mRNAs enriched in rare codons, using HIV-1 genes as a model. We found that, in a variety of cell types, including primary immune cells, SLFN14 regulates the expression of HIV-1 and non-viral genes based on their codon adaptation index, a measurement of the synonymous codon usage bias; consequently, SLFN14 inhibits the replication of HIV-1. The potent inhibitory effect of SLFN14 on the expression of the rare codon-rich transcript HIV-1 Gag was minimized by codon optimization. Mechanistically, we found that the endoribonuclease activity of SLFN14 is required, and that ribosomal RNA degradation is involved. Therefore, we propose that SLFN14 impairs the expression of HIV-1 transcripts rich in rare codons, in a catalytic-dependent manner.


Subject(s)
Codon Usage , HIV-1 , Virus Replication , Humans , Codon/genetics , Gene Expression Regulation, Viral , HEK293 Cells , HIV Infections/virology , HIV Infections/genetics , HIV-1/genetics , HIV-1/physiology , Protein Biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Cell Line, Tumor
6.
Viruses ; 16(4)2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38675950

ABSTRACT

Hepatitis B virus (HBV) is the etiologic agent of chronic hepatitis B, which puts at least 300 million patients at risk of developing fibrosis, cirrhosis, and hepatocellular carcinoma. HBV is a partially double-stranded DNA virus of the Hepadnaviridae family. While HBV was discovered more than 50 years ago, many aspects of its replicative cycle remain incompletely understood. Central to HBV persistence is the formation of covalently closed circular DNA (cccDNA) from the incoming relaxed circular DNA (rcDNA) genome. cccDNA persists as a chromatinized minichromosome and is the major template for HBV gene transcription. Here, we review how cccDNA and the viral minichromosome are formed and how viral gene transcription is regulated and highlight open questions in this area of research.


Subject(s)
DNA, Circular , DNA, Viral , Hepatitis B virus , Virus Replication , Hepatitis B virus/genetics , Hepatitis B virus/physiology , DNA, Circular/genetics , Humans , DNA, Viral/genetics , Viral Transcription/genetics , Gene Expression Regulation, Viral , Transcription, Genetic , Genome, Viral , Hepatitis B, Chronic/virology , Hepatitis B/virology , DNA Replication
7.
J Virol ; 98(5): e0003224, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38651900

ABSTRACT

Critical stages of lytic herpes simplex virus type 1 (HSV-1) replication are marked by the sequential expression of immediate early (IE) to early (E), then late (L) viral genes. HSV-1 can also persist in neuronal cells via a non-replicative, transcriptionally repressed infection called latency. The regulation of lytic and latent transcriptional profiles is critical to HSV-1 pathogenesis and persistence. We sought a fluorescence-based approach to observe the outcome of neuronal HSV-1 infection at the single-cell level. To achieve this goal, we constructed and characterized a novel HSV-1 recombinant that enables discrimination between lytic and latent infection. The dual reporter HSV-1 encodes a human cytomegalovirus-immediate early (hCMV-IE) promoter-driven enhanced yellow fluorescent protein (eYFP) to visualize the establishment of infection and an endogenous mCherry-VP26 fusion to report lytic replication. We confirmed that viral gene expression, replication, and spread of infection are not altered by the incorporation of the fluorescent reporters, and fluorescent protein (FP) detection virtuously reports the progression of lytic replication. We demonstrate that the outcome of HSV-1 infection of compartmentalized primary neurons is determined by viral inoculating dose: high-dose axonal inoculation proceeds to lytic replication, whereas low-dose axonal inoculation establishes a latent HSV-1 infection. Interfering with low-dose axonal inoculation via small molecule drugs reports divergent phenotypes of eYFP and mCherry reporter detection, correlating with altered states of viral gene expression. We report that the transcriptional state of neuronal HSV-1 infection is variable in response to changes in the intracellular neuronal environment.IMPORTANCEHerpes simplex virus type 1 (HSV-1) is a prevalent human pathogen that infects approximately 67% of the global human population. HSV-1 invades the peripheral nervous system, where latent HSV-1 infection persists within the host for life. Immunological evasion, viral persistence, and herpetic pathologies are determined by the regulation of HSV-1 gene expression. Studying HSV-1 gene expression during neuronal infection is challenging but essential for the development of antiviral therapeutics and interventions. We used a recombinant HSV-1 to evaluate viral gene expression during infection of primary neurons. Manipulation of cell signaling pathways impacts the establishment and transcriptional state of HSV-1 latency in neurons. The work here provides critical insight into the cellular and viral factors contributing to the establishment of latent HSV-1 infection.


Subject(s)
Herpes Simplex , Herpesvirus 1, Human , Luminescent Proteins , Neurons , Virus Replication , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/physiology , Neurons/virology , Neurons/metabolism , Humans , Animals , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Herpes Simplex/virology , Genes, Reporter , Virus Latency/genetics , Gene Expression Regulation, Viral , Chlorocebus aethiops , Vero Cells , Cytomegalovirus/genetics , Cytomegalovirus/physiology
8.
Viruses ; 16(4)2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38675871

ABSTRACT

The post-transcriptional regulatory element (PRE) is present in all HBV mRNAs and plays a major role in their stability, nuclear export, and enhancement of viral gene expression. Understanding PRE's structure, function, and mode of action is essential to leverage its potential as a therapeutic target. A wide range of PRE-based reagents and tools have been developed and assessed in preclinical and clinical settings for therapeutic and biotechnology applications. This manuscript aims to provide a systematic review of the characteristics and mechanism of action of PRE, as well as elucidating its current applications in basic and clinical research. Finally, we discuss the promising opportunities that PRE may provide to antiviral development, viral biology, and potentially beyond.


Subject(s)
Hepatitis B virus , Hepatitis B , RNA, Viral , Hepatitis B virus/genetics , Hepatitis B virus/physiology , Humans , Hepatitis B/virology , Hepatitis B/drug therapy , RNA, Viral/genetics , RNA, Viral/metabolism , Antiviral Agents/therapeutic use , Antiviral Agents/pharmacology , Gene Expression Regulation, Viral , RNA, Messenger/genetics , RNA Processing, Post-Transcriptional , Animals
9.
Viruses ; 16(4)2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38675925

ABSTRACT

The interferon-induced host cell protein Shiftless (SFL) inhibits -1 programmed ribosomal frameshifting (-1PRF) required for the expression of HIV-1 Gal-Pol and the formation of infectious HIV-1 particles. However, the specific regions in SFL required for antiviral activity and the mechanism by which SFL inhibits -1PRF remain unclear. Employing alanine scanning mutagenesis, we found that basic amino acids in the predicted zinc ribbon motif of SFL are essential for the suppression of Gag-Pol expression but dispensable for anti-HIV-1 activity. We have shown that SFL inhibits the expression of the murine leukemia virus (MLV) Gag-Pol polyprotein and the formation of infectious MLV particles, although Gag-Pol expression of MLV is independent of -1PRF but requires readthrough of a stop codon. These findings indicate that SFL might inhibit HIV-1 infection by more than one mechanism and that SFL might target programmed translational readthrough as well as -1PRF signals, both of which are regulated by mRNA secondary structure elements.


Subject(s)
Fusion Proteins, gag-pol , HIV Infections , HIV-1 , HIV-1/genetics , HIV-1/physiology , Humans , HIV Infections/virology , HIV Infections/genetics , HIV Infections/metabolism , Fusion Proteins, gag-pol/genetics , Fusion Proteins, gag-pol/metabolism , Leukemia Virus, Murine/genetics , Leukemia Virus, Murine/physiology , Frameshifting, Ribosomal , Virus Replication , HEK293 Cells , Gene Expression Regulation, Viral
10.
Viruses ; 16(4)2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38675926

ABSTRACT

The transcription and replication of the Newcastle disease virus (NDV) strictly rely on the viral ribonucleoprotein (RNP) complex, which is composed of viral NP, P, L and RNA. However, it is not known whether other viral non-RNP proteins participate in this process for viral self-regulation. In this study, we used a minigenome (MG) system to identify the regulatory role of the viral non-RNP proteins V, M, W, F and HN. Among them, V significantly reduced MG-encoded reporter activity compared with the other proteins and inhibited the synthesis of viral mRNA and cRNA. Further, V interacted with NP. A mutation in residue W195 of V diminished V-NP interaction and inhibited inclusion body (IB) formation in NP-P-L-cotransfected cells. Furthermore, a reverse-genetics system for the highly virulent strain F48E9 was established. The mutant rF48E9-VW195R increased viral replication and apparently enhanced IB formation. In vivo experiments demonstrated that rF48E9-VW195R decreased virulence and retarded time of death. Overall, the results indicate that the V-NP interaction of the W195 mutant V decreased, which regulated viral RNA synthesis, IB formation, viral replication and pathogenicity. This study provides insight into the self-regulation of non-RNP proteins in paramyxoviruses.


Subject(s)
Newcastle disease virus , Viral Proteins , Virus Replication , Newcastle disease virus/genetics , Newcastle disease virus/physiology , Newcastle disease virus/metabolism , Animals , Viral Proteins/metabolism , Viral Proteins/genetics , Nucleoproteins/metabolism , Nucleoproteins/genetics , Newcastle Disease/virology , Newcastle Disease/metabolism , Cell Line , Gene Expression Regulation, Viral , RNA, Viral/genetics , RNA, Viral/metabolism , Chickens , Virulence , Protein Binding , Mutation
11.
Viruses ; 16(4)2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38675956

ABSTRACT

Chronic hepatitis B (CHB) virus infection is a major public health burden and the leading cause of hepatocellular carcinoma. Despite the efficacy of current treatments, hepatitis B virus (HBV) cannot be fully eradicated due to the persistence of its minichromosome, or covalently closed circular DNA (cccDNA). The HBV community is investing large human and financial resources to develop new therapeutic strategies that either silence or ideally degrade cccDNA, to cure HBV completely or functionally. cccDNA transcription is considered to be the key step for HBV replication. Transcription not only influences the levels of viral RNA produced, but also directly impacts their quality, generating multiple variants. Growing evidence advocates for the role of the co-transcriptional regulation of HBV RNAs during CHB and viral replication, paving the way for the development of novel therapies targeting these processes. This review focuses on the mechanisms controlling the different co-transcriptional processes that HBV RNAs undergo, and their contribution to both viral replication and HBV-induced liver pathogenesis.


Subject(s)
Gene Expression Regulation, Viral , Hepatitis B virus , RNA, Viral , Virus Replication , Hepatitis B virus/genetics , Hepatitis B virus/physiology , Humans , RNA, Viral/genetics , Hepatitis B, Chronic/virology , DNA, Circular/genetics , DNA, Circular/metabolism , Transcription, Genetic , Animals , DNA, Viral/genetics
12.
PLoS Pathog ; 20(4): e1012172, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38662769

ABSTRACT

The implementation of antiretroviral therapy (ART) has effectively restricted the transmission of Human Immunodeficiency Virus (HIV) and improved overall clinical outcomes. However, a complete cure for HIV remains out of reach, as the virus persists in a stable pool of infected cell reservoir that is resistant to therapy and thus a main barrier towards complete elimination of viral infection. While the mechanisms by which host proteins govern viral gene expression and latency are well-studied, the emerging regulatory functions of non-coding RNAs (ncRNA) in the context of T cell activation, HIV gene expression and viral latency have not yet been thoroughly explored. Here, we report the identification of the Cytoskeleton Regulator (CYTOR) long non-coding RNA (lncRNA) as an activator of HIV gene expression that is upregulated following T cell stimulation. Functional studies show that CYTOR suppresses viral latency by directly binding to the HIV promoter and associating with the cellular positive transcription elongation factor (P-TEFb) to activate viral gene expression. CYTOR also plays a global role in regulating cellular gene expression, including those involved in controlling actin dynamics. Depletion of CYTOR expression reduces cytoplasmic actin polymerization in response to T cell activation. In addition, treating HIV-infected cells with pharmacological inhibitors of actin polymerization reduces HIV gene expression. We conclude that both direct and indirect effects of CYTOR regulate HIV gene expression.


Subject(s)
Gene Expression Regulation, Viral , HIV Infections , HIV-1 , RNA, Long Noncoding , Virus Latency , Humans , RNA, Long Noncoding/genetics , HIV Infections/virology , HIV Infections/genetics , HIV-1/genetics , HIV-1/physiology , Promoter Regions, Genetic , Lymphocyte Activation , Jurkat Cells
13.
PLoS Pathog ; 20(4): e1011939, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38683861

ABSTRACT

Epstein-Barr virus (EBV) persistently infects 95% of adults worldwide and is associated with multiple human lymphomas that express characteristic EBV latency programs used by the virus to navigate the B-cell compartment. Upon primary infection, the EBV latency III program, comprised of six Epstein-Barr Nuclear Antigens (EBNA) and two Latent Membrane Protein (LMP) antigens, drives infected B-cells into germinal center (GC). By incompletely understood mechanisms, GC microenvironmental cues trigger the EBV genome to switch to the latency II program, comprised of EBNA1, LMP1 and LMP2A and observed in GC-derived Hodgkin lymphoma. To gain insights into pathways and epigenetic mechanisms that control EBV latency reprogramming as EBV-infected B-cells encounter microenvironmental cues, we characterized GC cytokine effects on EBV latency protein expression and on the EBV epigenome. We confirmed and extended prior studies highlighting GC cytokine effects in support of the latency II transition. The T-follicular helper cytokine interleukin 21 (IL-21), which is a major regulator of GC responses, and to a lesser extent IL-4 and IL-10, hyper-induced LMP1 expression, while repressing EBNA expression. However, follicular dendritic cell cytokines including IL-15 and IL-27 downmodulate EBNA but not LMP1 expression. CRISPR editing highlighted that STAT3 and STAT5 were necessary for cytokine mediated EBNA silencing via epigenetic effects at the EBV genomic C promoter. By contrast, STAT3 was instead necessary for LMP1 promoter epigenetic remodeling, including gain of activating histone chromatin marks and loss of repressive polycomb repressive complex silencing marks. Thus, EBV has evolved to coopt STAT signaling to oppositely regulate the epigenetic status of key viral genomic promoters in response to GC cytokine cues.


Subject(s)
Epigenesis, Genetic , Epstein-Barr Virus Infections , Gene Expression Regulation, Viral , Germinal Center , Herpesvirus 4, Human , Virus Latency , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/physiology , Humans , Germinal Center/immunology , Germinal Center/virology , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/immunology , Cytokines/metabolism , B-Lymphocytes/virology , B-Lymphocytes/metabolism
14.
PLoS Pathog ; 20(3): e1011998, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38530845

ABSTRACT

Epstein-Barr virus (EBV) contributes to ~1% of all human cancers including several B-cell neoplasms. A characteristic feature of EBV life cycle is its ability to transform metabolically quiescent B-lymphocytes into hyperproliferating B-cell blasts with the establishment of viral latency, while intermittent lytic cycle induction is necessary for the production of progeny virus. Our RNA-Seq analyses of both latently infected naïve B-lymphocytes and transformed B-lymphocytes upon lytic cycle replication indicate a contrasting expression pattern of a membrane-associated carbonic anhydrase isoform CA9, an essential component for maintaining cell acid-base homeostasis. We show that while CA9 expression is transcriptionally activated during latent infection model, lytic cycle replication restrains its expression. Pharmacological inhibition of CA-activity using specific inhibitors retards EBV induced B-cell transformation, inhibits B-cells outgrowth and colony formation ability of transformed B-lymphocytes through lowering the intracellular pH, induction of cell apoptosis and facilitating degradation of CA9 transcripts. Reanalyses of ChIP-Seq data along with utilization of EBNA2 knockout virus, ectopic expression of EBNA2 and sh-RNA mediated knockdown of CA9 expression we further demonstrate that EBNA2 mediated CA9 transcriptional activation is essential for EBV latently infected B-cell survival. In contrast, during lytic cycle reactivation CA9 expression is transcriptionally suppressed by the key EBV lytic cycle transactivator, BZLF1 through its transactivation domain. Overall, our study highlights the dynamic alterations of CA9 expression and its activity in regulating pH homeostasis act as one of the major drivers for EBV induced B-cell transformation and subsequent B-cell lymphomagenesis.


Subject(s)
Epstein-Barr Virus Infections , Herpesvirus 4, Human , Humans , Herpesvirus 4, Human/physiology , Epstein-Barr Virus Infections/genetics , B-Lymphocytes , Virus Latency , Trans-Activators/genetics , Virus Activation , Gene Expression Regulation, Viral
15.
J Virol ; 98(4): e0024224, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38446633

ABSTRACT

Viral genomes frequently harbor overlapping genes, complicating the development of virus-vectored vaccines and gene therapies. This study introduces a novel conditional splicing system to precisely control the expression of such overlapping genes through recombinase-mediated conditional splicing. We refined site-specific recombinase (SSR) conditional splicing systems and explored their mechanisms. The systems demonstrated exceptional inducibility (116,700-fold increase) with negligible background expression, facilitating the conditional expression of overlapping genes in adenovirus-associated virus (AAV) and human immunodeficiency virus type 1. Notably, this approach enabled the establishment of stable AAV producer cell lines, encapsulating all necessary packaging genes. Our findings underscore the potential of the SSR-conditional splicing system to significantly advance vector engineering, enhancing the efficacy and scalability of viral-vector-based therapies and vaccines. IMPORTANCE: Regulating overlapping genes is vital for gene therapy and vaccine development using viral vectors. The regulation of overlapping genes presents challenges, including cytotoxicity and impacts on vector capacity and genome stability, which restrict stable packaging cell line development and broad application. To address these challenges, we present a "loxp-splice-loxp"-based conditional splicing system, offering a novel solution for conditional expression of overlapping genes and stable cell line establishment. This system may also regulate other cytotoxic genes, representing a significant advancement in cell engineering and gene therapy as well as biomass production.


Subject(s)
Dependovirus , Genes, Overlapping , Genes, Viral , Genetic Engineering , HIV-1 , RNA Splicing , Humans , Cell Line , Dependovirus/genetics , DNA Nucleotidyltransferases/genetics , DNA Nucleotidyltransferases/metabolism , Gene Expression Regulation, Viral , Genes, Overlapping/genetics , Genes, Viral/genetics , Genetic Engineering/methods , Genetic Therapy/methods , Genetic Vectors/genetics , HIV-1/genetics , RNA Splicing/genetics , Vaccines/biosynthesis , Vaccines/genetics , Viral Genome Packaging/genetics
16.
J Virol ; 98(4): e0201523, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38451083

ABSTRACT

Herpes simplex virus 1 (HSV-1) transcription is restricted in latently infected neurons and the genomes are in mostly silenced chromatin, whereas all viral genes are transcribed in lytically infected cells, in which the genomes are dynamically chromatinized. Epigenetic regulation modulates HSV-1 transcription during lytic, latent, and reactivating infections but the precise mechanisms are not fully defined. Nucleosomes are dynamic: they slide, breathe, assemble, and disassemble. We and others have proposed that the most dynamic HSV-1 chromatin is transcriptionally competent, whereas the least dynamic is silenced. However, the mechanisms yielding the unusually dynamic viral chromatin remain unknown. Histone variants affect nucleosome dynamics. The dynamics of H2A, H2A.X, and macroH2A were enhanced in infected cells, whereas those of H2A.B were uniquely decreased. We constructed stably transduced cells expressing tagged histone H2A, H2A.B, macroH2A, or H2B, which assembles the H2A/H2B nucleosome dimers with all H2A variants. All H2A variants, as well as ectopic and endogenous H2B were assembled into HSV-1 chromatin evenly throughout the genome but canonical H2A was relatively depleted whereas H2A.B was enriched, particularly in the most dynamic viral chromatin. When viral transcription and DNA replication were restricted, H2A.B became as depleted from the viral chromatin through the entire genome as H2A. We propose that lytic HSV-1 nucleosomes are enriched in the dynamic variant H2A.B/H2B dimers to promote HSV-1 chromatin dynamics and transcriptional competency and conclude that the dynamics of HSV-1 chromatin are determined in part by the H2A variants. IMPORTANCE: Herpes simplex virus 1 (HSV-1) transcription is epigenetically regulated during latent and lytic infections, and epigenetic inhibitors have been proposed as potential antiviral drugs to modulate latency and reactivation. However, the detailed epigenetic mechanisms of regulation of HSV-1 transcription have not been fully characterized and may differ from those regulating cellular transcription. Whereas lytic HSV-1 chromatin is unusually dynamic, latent silenced HSV-1 chromatin is not. The mechanisms resulting in the unique dynamics of the lytic chromatin remain unknown. Here we identify the enrichment of the highly dynamic histone 2A variant H2A in the most dynamic viral chromatin, which provides a mechanistic understanding of its unique dynamics. Future work to identify the mechanisms of enrichment in H2A.B on the viral chromatin may identify novel druggable epigenetic regulators that modulate HSV-1 latency and reactivation.


Subject(s)
Chromatin , Epigenesis, Genetic , Gene Expression Regulation, Viral , Herpesvirus 1, Human , Histones , Viral Transcription , Virus Replication , Chromatin/genetics , Chromatin/metabolism , Gene Silencing , Genetic Variation , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/growth & development , Herpesvirus 1, Human/physiology , Histones/genetics , Histones/metabolism , Nucleosomes/genetics , Nucleosomes/metabolism , Virus Activation , Virus Latency , Humans , Animals , Vero Cells , HEK293 Cells
17.
J Virol ; 98(3): e0150223, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38315015

ABSTRACT

Capsid assembly is critical in the hepatitis B virus (HBV) life cycle, mediated by the viral core protein. Capsid assembly is the target for new anti-viral therapeutics known as capsid assembly modulators (CAMs) of which the CAM-aberrant (CAM-A) class induces aberrant shaped core protein structures and leads to hepatocyte cell death. This study aimed to identify the mechanism of action of CAM-A modulators leading to HBV-infected hepatocyte elimination where CAM-A-mediated hepatitis B surface antigen (HBsAg) reduction was evaluated in a stable HBV replicating cell line and in AAV-HBV-transduced C57BL/6, C57BL/6 SCID, and HBV-infected chimeric mice with humanized livers. Results showed that in vivo treatment with CAM-A modulators induced pronounced reductions in hepatitis B e antigen (HBeAg) and HBsAg, associated with a transient alanine amino transferase (ALT) increase. Both HBsAg and HBeAg reductions and ALT increase were delayed in C57BL/6 SCID and chimeric mice, suggesting that adaptive immune responses may indirectly contribute. However, CD8+ T cell depletion in transduced wild-type mice did not impact antigen reduction, indicating that CD8+ T cell responses are not essential. Transient ALT elevation in AAV-HBV-transduced mice coincided with a transient increase in endoplasmic reticulum stress and apoptosis markers, followed by detection of a proliferation marker. Microarray data revealed antigen presentation pathway (major histocompatibility complex class I molecules) upregulation, overlapping with the apoptosis. Combination treatment with HBV-specific siRNA demonstrated that CAM-A-mediated HBsAg reduction is dependent on de novo core protein translation. To conclude, CAM-A treatment eradicates HBV-infected hepatocytes with high core protein levels through the induction of apoptosis, which can be a promising approach as part of a regimen to achieve functional cure. IMPORTANCE: Treatment with hepatitis B virus (HBV) capsid assembly modulators that induce the formation of aberrant HBV core protein structures (CAM-A) leads to programmed cell death, apoptosis, of HBV-infected hepatocytes and subsequent reduction of HBV antigens, which differentiates CAM-A from other CAMs. The effect is dependent on the de novo synthesis and high levels of core protein.


Subject(s)
Antiviral Agents , Apoptosis , Gene Expression Regulation, Viral , Hepatitis B Core Antigens , Hepatitis B virus , Hepatocytes , Protein Biosynthesis , Animals , Mice , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Apoptosis/drug effects , Capsid/chemistry , Capsid/classification , Capsid/drug effects , Capsid/metabolism , Capsid Proteins/metabolism , Hepatitis B/drug therapy , Hepatitis B/immunology , Hepatitis B/metabolism , Hepatitis B/virology , Hepatitis B Core Antigens/biosynthesis , Hepatitis B Core Antigens/metabolism , Hepatitis B e Antigens/metabolism , Hepatitis B Surface Antigens/metabolism , Hepatitis B virus/growth & development , Hepatitis B virus/immunology , Hepatitis B virus/metabolism , Hepatitis B virus/pathogenicity , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Hepatocytes/virology , Mice, Inbred C57BL , Mice, SCID , Virus Replication , Cell Line , CD8-Positive T-Lymphocytes/immunology , Antigen Presentation
18.
J Virol ; 98(3): e0151523, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38323812

ABSTRACT

Adeno-associated virus (AAV) requires co-infection with helper virus for efficient replication. We previously reported that Human Bocavirus 1 (HBoV1) genes, including NP1, NS2, and BocaSR, were critical for AAV2 replication. Here, we first demonstrate the essential roles of the NP1 protein in AAV2 DNA replication and protein expression. We show that NP1 binds to single-strand DNA (ssDNA) at least 30 nucleotides (nt) in length in a sequence-independent manner. Furthermore, NP1 colocalized with the BrdU-labeled AAV2 DNA replication center, and the loss of the ssDNA-binding ability of NP1 by site-directed mutation completely abolished AAV2 DNA replication. We used affinity-tagged NP1 protein to identify host cellular proteins associated with NP1 in cells cotransfected with the HBoV1 helper genes and AAV2 duplex genome. Of the identified proteins, we demonstrate that NP1 directly binds to the DBD-F domain of the RPA70 subunit with a high affinity through the residues 101-121. By reconstituting the heterotrimer protein RPA in vitro using gel filtration, we demonstrate that NP1 physically associates with RPA to form a heterologous complex characterized by typical fast-on/fast-off kinetics. Following a dominant-negative strategy, we found that NP1-RPA complex mainly plays a role in expressing AAV2 capsid protein by enhancing the transcriptional activity of the p40 promoter. Our study revealed a novel mechanism by which HBoV1 NP1 protein supports AAV2 DNA replication and capsid protein expression through its ssDNA-binding ability and direct interaction with RPA, respectively.IMPORTANCERecombinant adeno-associated virus (rAAV) vectors have been extensively used in clinical gene therapy strategies. However, a limitation of these gene therapy strategies is the efficient production of the required vectors, as AAV alone is replication-deficient in the host cells. HBoV1 provides the simplest AAV2 helper genes consisting of NP1, NS2, and BocaSR. An important question regarding the helper function of HBoV1 is whether it provides any direct function that supports AAV2 DNA replication and protein expression. Also of interest is how HBoV1 interplays with potential host factors to constitute a permissive environment for AAV2 replication. Our studies revealed that the multifunctional protein NP1 plays important roles in AAV2 DNA replication via its sequence-independent ssDNA-binding ability and in regulating AAV2 capsid protein expression by physically interacting with host protein RPA. Our findings present theoretical guidance for the future application of the HBoV1 helper genes in the rAAV vector production.


Subject(s)
Capsid Proteins , Capsid , DNA, Single-Stranded , DNA, Viral , DNA-Binding Proteins , Dependovirus , Human bocavirus , Viral Proteins , Humans , Capsid/metabolism , Capsid Proteins/biosynthesis , Capsid Proteins/chemistry , Capsid Proteins/genetics , Capsid Proteins/metabolism , Dependovirus/genetics , Dependovirus/growth & development , Dependovirus/metabolism , DNA, Single-Stranded/biosynthesis , DNA, Single-Stranded/metabolism , DNA, Viral/biosynthesis , DNA, Viral/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Viral , Human bocavirus/genetics , Human bocavirus/metabolism , Kinetics , Mutagenesis, Site-Directed , Mutation , Promoter Regions, Genetic , Protein Binding , Protein Domains , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication
19.
mBio ; 15(3): e0347923, 2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38349188

ABSTRACT

Productive replication of herpes simplex virus (HSV) relies upon a well-ordered transcriptional cascade flowing from immediate-early (IE) to early (E) to late (L) gene products. While several virus-encoded transcriptional activators are involved in this process, IE and E gene promoters also contain multiple binding sites for the ubiquitously expressed cellular transcription factor Sp1. Sp1 has been previously implicated in activating HSV-1 gene transcription downstream of these sites, but why Sp1-binding sites are maintained in the promoters of genes activated by virus-encoded activators remains unclear. We hypothesized that Sp1 enables continued HSV-1 transcription and replication when viral transactivators are limited. We used a depletion-based approach in human foreskin fibroblasts to investigate the specific contribution of Sp1 to the initiation and progression of the HSV-1 lytic gene cascade. We found that Sp1 increased viral transcript levels, protein expression, and replication following infection with VP16- or ICP0-deficient viruses but had little to no effect on rescued viruses or during wild-type (WT) HSV-1 infection. Moreover, Sp1 promoted WT virus transcription and replication following interferon treatment of fibroblasts and thus may contribute to viral immune evasion. Interestingly, we observed reduced expression of Sp1 and Sp1-family transcription factors in differentiated sensory neurons compared to undifferentiated cells, suggesting that reduced Sp1 levels may also contribute to HSV-1 latent infection. Overall, these findings indicate that Sp1 can promote HSV-1 gene expression in the absence of key viral transactivators; thus, HSV-1 may use Sp1 to maintain its gene expression and replication under adverse conditions.IMPORTANCEHerpes simplex virus (HSV) is a common human pathogen that actively replicates in the epithelia but can persist for the lifetime of the infected host via a stable, latent infection in neurons. A key feature of the HSV replication cycle is a complex transcriptional program in which virus and host-cell factors coordinate to regulate expression of the viral gene products necessary for continued viral replication. Multiple binding sites for the cellular transcription factor Sp1 are located in the promoters of HSV-1 genes, but how Sp1 binding contributes to transcription and replication of wild-type virus is not fully understood. In this study, we identified a specific role for Sp1 in maintaining HSV-1 gene transcription under adverse conditions, as when virus-encoded transcriptional activators were absent or limited. Preservation of Sp1-binding sites in HSV-1 gene promoters may thus benefit the virus as it navigates diverse cell types and host-cell conditions during infection.


Subject(s)
Herpes Simplex , Herpesviridae Infections , Herpesvirus 1, Human , Immediate-Early Proteins , Latent Infection , Humans , Herpesvirus 1, Human/physiology , Trans-Activators/genetics , Immediate-Early Proteins/genetics , Transcription Factors/metabolism , Virus Replication , Gene Expression , Gene Expression Regulation, Viral
20.
Proc Natl Acad Sci U S A ; 121(10): e2315860121, 2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38408244

ABSTRACT

Human cytomegalovirus (HCMV) is a prevalent pathogen that establishes life-long latent infection in hematopoietic cells. While this infection is usually asymptomatic, immune dysregulation leads to viral reactivation, which can cause significant morbidity and mortality. However, the mechanisms underpinning reactivation remain incompletely understood. The HCMV major immediate early promoter (MIEP)/enhancer is a key factor in this process, as its transactivation from a repressed to active state helps drive viral gene transcription necessary for reactivation from latency. Numerous host transcription factors bind the MIE locus and recruit repressive chromatin modifiers, thus impeding virus reactivation. One such factor is CCCTC-binding protein (CTCF), a highly conserved host zinc finger protein that mediates chromatin conformation and nuclear architecture. However, the mechanisms by which CTCF contributes to HCMV latency were previously unexplored. Here, we confirm that CTCF binds two convergent sites within the MIE locus during latency in primary CD14+ monocytes, and following cellular differentiation, CTCF association is lost as the virus reactivates. While mutation of the MIE enhancer CTCF binding site does not impact viral lytic growth in fibroblasts, this mutant virus fails to maintain latency in myeloid cells. Furthermore, we show the two convergent CTCF binding sites allow looping to occur across the MIEP, supporting transcriptional repression during latency. Indeed, looping between the two sites diminishes during virus reactivation, concurrent with activation of MIE transcription. Taken together, our data reveal that three-dimensional chromatin looping aids in the regulation of HCMV latency and provides insight into promoter/enhancer regulation that may prove broadly applicable across biological systems.


Subject(s)
Cytomegalovirus Infections , Cytomegalovirus , Humans , Chromatin/genetics , Cytomegalovirus/genetics , Cytomegalovirus Infections/genetics , Gene Expression Regulation, Viral , Promoter Regions, Genetic , Virus Activation/genetics , Virus Latency/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...